Next Article in Journal
Csf1 Deficiency Dysregulates Glial Responses to Demyelination and Disturbs CNS White Matter Remyelination
Next Article in Special Issue
DAF-16/FoxO in Caenorhabditis elegans and Its Role in Metabolic Remodeling
Previous Article in Journal
Extracellular Vesicles from Mesenchymal Stem Cells as Novel Treatments for Musculoskeletal Diseases
Previous Article in Special Issue
Expression of Adenoviral E1A in Transformed Cells as an Additional Factor of HDACi-Dependent FoxO Regulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Signaling Network of Forkhead Family of Transcription Factors (FOXO) in Dietary Restriction

1
Centre of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
2
Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China
3
School of Medical Science, Jinan University, Guangzhou 510080, China
4
Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
5
School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2020, 9(1), 100; https://doi.org/10.3390/cells9010100
Submission received: 21 November 2019 / Revised: 25 December 2019 / Accepted: 29 December 2019 / Published: 31 December 2019
(This article belongs to the Special Issue The FoxO Transcription Factors and Metabolic Regulation)

Abstract

:
Dietary restriction (DR), which is defined as a reduction of particular or total nutrient intake without causing malnutrition, has been proved to be a robust way to extend both lifespan and health-span in various species from yeast to mammal. However, the molecular mechanisms by which DR confers benefits on longevity were not yet fully elucidated. The forkhead box O transcription factors (FOXOs), identified as downstream regulators of the insulin/IGF-1 signaling pathway, control the expression of many genes regulating crucial biological processes such as metabolic homeostasis, redox balance, stress response and cell viability and proliferation. The activity of FOXOs is also mediated by AMP-activated protein kinase (AMPK), sirtuins and the mammalian target of rapamycin (mTOR). Therefore, the FOXO-related pathways form a complex network critical for coordinating a response to environmental fluctuations in order to maintain cellular homeostasis and to support physiological aging. In this review, we will focus on the role of FOXOs in different DR interventions. As different DR regimens or calorie (energy) restriction mimetics (CRMs) can elicit both distinct and overlapped DR-related signaling pathways, the benefits of DR may be maximized by combining diverse forms of interventions. In addition, a better understanding of the precise role of FOXOs in different mechanistic aspects of DR response would provide clear cellular and molecular insights on DR-induced increase of lifespan and health-span.

1. Dietary Restriction and FOXOs

Dietary restriction (DR), which is defined as a reduction of particular or total nutrient intake without causing malnutrition, has been proved to be a robust method to decelerate the aging process. Besides extended lifespan, animals subjected to DR also display other phenotypes including increased stress resistance, increased insulin sensitivity, improved mitochondrial health, decreased lipid accumulation and reduced body size. For example, mice under DR survive better than ad libitum fed animals under paraquat-induced oxidative stress [1]. Both short- and long-term DR mitigate ischemia reperfusion injury in the brain, heart, liver, and kidney by activating autophagy, reducing the inflammatory response and enhancing antioxidant activity [2,3,4,5,6]. In humans, DR improves several biomarkers of longevity and reduces the risk of developing diabetes, cardiovascular disease and cancer [7,8].
Forkhead box O transcription factors (FOXO) transcription factors have long been considered as a key regulator of aging. The mammalian FOXO family includes four proteins: FOXO1, FOXO3 (or FOXO3a), FOXO4 and FOXO6 [9]. Invertebrates express a single FOXO gene, for example, daf (abnormal dauer formation)-16 in Caenorhabditis elegans [10] and dFOXO in Drosophila [11]. Albeit, FOXOs have distinct expression patterns and functions, they recognize the same consensus sequence TTGTTTAC [12]. In response to changing environment or internal stimuli, FOXOs bind to the promoter of many target genes that are involved in a variety of crucial biological processes such as stress resistance, metabolism, proliferation, cell cycle arrest and autophagy [13,14,15]. FOXOs have been shown to play a key role in DR response. In yeasts, DR failed to extend the chronological lifespan of mutant with deletion of both FKH1 and FKH2, two Forkhead box orthologs which are considered to be evolutionarily conserved with higher eukaryotic FOXO proteins [16]. In rat skeletal muscles, every other day feeding resulted in significantly elevated gene expression of FoxO1 and 4, whereas FoxO3 levels were not statistically significantly elevated [17]. In mice, DR up-regulated mRNA expression levels of FoxO1 in the liver and skeletal muscle, increased the levels of FoxO4 mRNA in the skeletal muscle and adipose tissue, whereas no effect was observed in Foxo3 mRNA levels in these tissues [18]. In addition to the direct DR-induced increase in FOXO proteins expression, DR also alters expression a variety of FOXO target genes, such as heme oxygenase-1 (HO-1), growth arrest and DNA-damage-inducible, alpha (GADD45a), manganese superoxide dismutase (MnSOD), catalase, cyclin-dependent kinase inhibitor 1A (p21) and cyclin-dependent kinase inhibitor 1B (p27) [4,18,19,20,21]. DR regimen did not extend the lifespan of both Foxo3-knockout heterozygous (+/−) and homozygous (−/−) mice, suggesting that FoxO3 is required for DR to extend lifespan [22]. The effect of DR on reducing incidence of tumor was abolished in FoxO1 knockout heterozygous (HT) mice, indicating that FoxO1 is involved in the anti-tumor effects of DR [18]. It was also reported that FoxO1-mediated antioxidant activity and autophagy are essential for the protective effect of DR against liver injury [4]. Moreover, a recent study reveals that FoxO1 also regulate the anti-inflammatory action of DR by interacting with PGC-1α (peroxisome proliferator-activated receptor gamma (PPAR-γ) coactivator 1-alpha) [23]. Interestingly, it was recently reported that in C. elegans, daf-16 could modulate immunity and promote longevity by inhibiting feeding behavior, which appears to mimic a DR-like situation [24]. The mechanism by which daf-16 regulates food intake is an interesting question deserving further investigation.

2. Signaling Networks of FOXO in DR: Insulin/IGF-1 Signaling Pathway

The activity of FOXOs is negatively regulated by IGF-1 (insulin-like growth factor 1) receptors. Briefly, ligand activation of IGF-1 receptor triggers the phosphorylation of PI3K (phosphoinositide 3-kinase) leading to activation of Akt (also called protein kinase B or PKB) by phosphorylation [25,26]. Akt phosphorylates FOXO proteins and promotes their binding to 14-3-3 proteins and nuclear exclusion [27,28]. FOXO protein levels in the cytosol are regulated by ubiquitin proteasome pathway (UPP) [29].
The insulin/IGF-1 signaling pathway is the first identified, longevity-regulating pathway. In C. elegans, mutation in daf-2, mammalian insulin/IGF-1 receptor homolog, results in a doubled lifespan [30]. Worm mutants carrying mutation in age-1, the C. elegans homolog of the PI3K, are also long-lived [31]. daf-16 is required for the extended lifespan of these mutants [32]. Interestingly, the extended lifespan of eat-2 mutant is not dependent on daf-16 and eat-2; daf-2 double mutants live longer than single mutants [33,34]. These findings suggest that DR and insulin/IGF-1 pathway differently act to extend lifespan in C. elegans. In the fruit fly Drosophila, DR did not further extend lifespan of a long-lived dwarf chico gene mutant, which carries a mutation in the insulin receptor (InR) substrate [35,36]. Conversely, another study found that although insulin-like peptide-5 (ilp5) was suppressed by DR, it was not required for DR to extend lifespan [37]. In addition, overexpression of dfoxo gene in the fat body increased the sensitivity to DR, but DR could still successfully extend lifespan in both dfoxo null mutant and wild-type. These findings indicate that although dfoxo modulates in part DR-induced lifespan extension, DR is utilizing additional signaling systems to regulate this process [37,38].
Although the Insulin/IGF-1 signaling pathway is a highly conserved from yeast to human, some major differences in its activity have been found in different species. For example, in mammals, but not in invertebrates, the activity of the insulin/IGF-1 pathway is regulated by growth hormone (GH), a peptide secreted by pituitary [39,40]. GH secretion declines with aging and its decreased secretion parallels the decrease in IGF-1 levels [39]. IGF-1 signaling is involved in the regulation of cell proliferation, stress resistance, apoptosis and tumorigenesis, and decreased IGF-1 level is proved to be lifespan beneficial. Studies in dogs have proved that animals with lower circulating IGF-1 levels tend to have smaller body size and longer lifespan [41,42]. A basic aging theory claims that GH decreases lifespan and that lifespan benefits of DR regimens are attributed to reduced growth stimulation [43,44]. DR has been shown to reduce IGF-1 levels in mice [45]. However, the effect of DR on IGF-1 signaling in humans appears to be more complex. A recent study showed that DR significantly decreased insulin and IGF-1 levels in humans, consistent with observations in rodents [46]. However, DR failed to decrease GH and IGF-1 levels in non-obese men and women [47]. Interestingly, another study demonstrated that although DR without malnutrition did not change IGF-1 levels in humans, a significant reduction was observed when protein intake was restricted [48]. These findings suggest that DR responses are different in rodents and humans, and therefore further studies are required to clarify the physiological and pathological metabolic effects of protein restriction.
Interventions to disrupt the GH/IGF-1 (somatotropic) axis have been shown to have pro-longevity effects in mice. For example, mice with deletion of insulin receptor substrate 1 (IRS1) are long-lived with delay age-related processes [49]. Knockout of the mouse growth hormone receptor/binding protein (GHR/BP) gene also results in an extended lifespan [44,50,51]. Interestingly, lifespan extension by suppression of the GH/IGF-1 system appears to require the complete absence of GH and IGF-1 signaling. It was reported that a single substitution of glycine with lysine at position 119 in bGH (bovine growth hormone) results in the production of a mutated protein that could act as a functional antagonist of GH [52]. Mice carrying such mutation have markedly reduced, low level of GH/IGF-1 signaling, and importantly, a normal lifespan [53]. GHR/BP −/− knock out mice also displayed DR-like phenotype such as GH resistance, decreased IGF-I levels and reduced body size [44,50,51]. In addition, DR did not alter metabolic parameters including oxygen consumption and respiratory quotient and failed to further extend the lifespan of GH receptor-deficient (GHRKO) mice, indicating that DR and reduced IGF-1/insulin signaling may act through the same mechanism to promote mice longevity [54,55]. Another study demonstrated that wild type mice subjected to DR and GHRKO mice have substantially different expression levels of genes related to insulin signaling, and this difference is tissue-specific [54]. Taken together, the effects of DR and reduced IGF-1/insulin signaling are similar but not identical, and this similarity could differ across species.

3. DR and AMPK

The AMP-activated protein kinase (AMPK) is an energy sensor that responds to cellular energy status and mediates metabolism. AMPK is activated by a high AMP/ATP ratio and therefore serves as a sensor of cellular “fuel deficiency”. AMPK activation is proposed to mediate some of the health-protective effects of long-term calorie restriction. AMP binds to AMPK and enables its activation by phosphorylation at Threonine (Thr)-172 by upstream kinases, including liver kinase B1 (LKB1) and calmodulin-dependent protein kinase (CaMKK) [56]. AMPK has been shown to directly regulate activity of FOXOs by phosphorylation [57]. AMPK phosphorylates FoxO3 at sites different from that was previously shown to be regulated by AKT [57]. Interestingly, AMPK appears to increase FoxO3 transcriptional activity without affecting its subcellular localization [57].
In C. elegans, aak-2, a homolog of the α-catalytic subunits of mammalian AMPK, regulates longevity in response to increased AMP/ATP ratio or decreased insulin-like signaling [58]. As reduced calorie intake has been shown to increase AMP/ATP ratio, it was hypothesized that aak-2 may mediate DR-induced lifespan extension [59]. Indeed, aak-2/eat-2 double mutant lives as long as eat-2 mutant, indicating that eat-2 deficiency-induced DR may bypass aak-2 to extend lifespan [60]. By contrary, a DR regimen in worms, called sDR, extended lifespan in an aak-2-dependent manner [61]. Worms expressing constitutively active AMPK exhibit increased resistance to oxidative stress, increased daf-16-dependent SOD-3 (Superoxide Dismutase 3) expression and prolonged lifespan [61]. In addition, daf-16 is necessary for both sDR and AMPK activation to extend lifespan and delay age-dependent fitness decline while daf-2 is not required [61]. Therefore, the AMPK/FOXO signaling pathway plays a key role in sDR-mediated lifespan extension. Another study indicated that AMPK and FOXO are necessary for lifespan extension induced by dilution of peptone, while are dispensable for longevity induced by bDR (a DR method achieved by diluting bacteria in liquid culture) and eat-2 (ad1116) mutation [62]. These findings propose that the role of the AMPK/FOXO pathway in DR could be different, depending on various DR regimens [63].

4. DR and SIRTUIN

The nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase silent information regulator 2 (Sir2) that targets both histone and non-histone proteins, including transcription factors involved in diverse processes such as stress resistance, cell differentiation, metabolism and aging is a metabolic regulator conserved across species [64,65]. It was reported that overexpressing Sir2 increases lifespan in both C. elegans and Drosophila [66,67]. In C. elegans, daf-16 is required for the lifespan extension of the sir-2.1 transgene, suggesting a possible interaction between Sir2 and daf-16 [66]. The transcriptional activity FOXO is regulated by reversible acetylation and deacetylation [68]. In response to oxidative stress stimuli, the cAMP response element-binding protein (CREB)-binding protein (CBP) acetylates FOXO and suppresses its transcriptional activity, by attenuating its DNA-binding ability and enhancing phosphorylation by Akt [68,69,70]. Sir2 binds FOXO and deacetylates it to promote FOXO-dependent gene transcription, including genes involved in oxidative stress resistance and glucose production [71]. Sir2 plays a key role in DR-mediated longevity. Animals subjecting to DR showed an increase in Sir2 protein levels [67,72,73]. In yeast, mutations in dSir2 or NPT1, the genes encoding NAD synthesis’ enzymes, blocked DR-induced increased longevity [74]. In Drosophila, DR failed to extend lifespan when dSir2 has no function or severely reduced function [67]. In C. elegans, SIR-2.1 binds DAF-16 in a 14-3-3-dependent manner to induce the transcriptional activation of DAF-16 [75]. The lifespan-extending effect in overexpressing sir-2.1 and eat-2 genes is not additive [76]. In addition, eat-2 mediated lifespan extension was suppressed in sir-2.1/eat-2 double mutant [77]. Therefore, sir-2.1 acts similarly to eat-2, to extend lifespan. However the regulation of life span is probably more complex since several other studies suggested that lifespan extension in response to DR can be independent of Sir2 [78,79].
Aging is associated with diminished SIRT1 expression and activity [80,81]. In mammals, DR induces the SIRT1, the closest homolog of C. elegans sir-2.1 and Drosophila dSir2, and promotes cell survival [82]. In animal models, stimulation of SIRT1 (either with agonists or NAD+ precursors) caused extension of the lifespan and protected against many aging-related diseases, including autoimmune diseases, cardiovascular diseases, neurodegenerative disorders and cancer [83,84,85,86,87,88,89]. In addition, DR induced an increase in physical activity was abolished in SIRT knockout mice [90]. Although daf-16 is required for the lifespan extension of the sir-2.1 overexpression [66], genetic analysis showed that the expression pattern of sir-2.1 and daf-16 is only partially overlapping [77]. UNC-13 regulates neurotransmitter release in the nervous system and serves as an upstream of daf-2, and the lifespan of mutants with some unc-13 alleles is more than doubled [77]. Interestingly, mutation in either sir-2.1 or daf-16 only partially suppressed the unc-13 induced lifespan extension in double mutants, whereas a triple mutant with unc-13; daf-16; sir-2.1 displayed a similar lifespan as the wild type, indicating the existence of separate functions for sir-2.1 and daf-16 in lifespan regulation [77]. The sirtuin activator resveratrol, also extends lifespan through a sir-2.1-dependent but daf-16-independent manner [62,91], which further supports the notion that DR and lifespan regulation via sir-2.1 and daf-16 are not in a simple signaling pathway. Taken together, Sir2 promotes longevity and mediates many of the beneficial effects of DR. Although Sir2/FOXO mediate DR response in some conditions, they can be replaceable under other conditions, and may also act in different signaling pathways to regulate DR response and longevity.

5. DR and mTOR

The mammalian target of rapamycin (mTOR), which was first identified as the target of rapamycin, is a conserved, nutrient sensing, serine/threonine protein kinase that regulates growth and metabolism in all eukaryotic cells. mTOR functions in two complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). These two complexes are consisting of both shared and distinct subunits. Both mTORC1 and mTORC2 contains mLST8/GβL and DEPTOR, while mTORC1 also contains PRAS40 and Raptor, and mTORC2-specific components include rapamycin-insensitive component of mTOR (Rictor), mSin1, and Protor-1/2 [40,92]. mTOR complexes regulate growth, metabolism, autophagy cell death and mRNA translation [93,94].
Inhibition of the TOR signaling pathway has been shown to mimic DR and extend lifespan in invertebrates and mammals. In yeast, inhibition of the TOR signaling pathway by gene mutations or pharmacological inhibition by methionine sulfoximine or rapamycin extended the life span [95]. Cells with suppressed TOR signaling displayed DR-like phenotypes such as an increase in accumulation of glycogen and increased stress resistance, indicating that mTOR signaling may mediate DR response [95]. However, there are also reports indicating that DR did not extend the lifespan of cells lacking TOR1, one of two TOR proteins expressed in yeast [96]. In Drosophila, inhibition of TOR signaling pathway by genetic manipulations, extended in a nutritional condition-dependent manner the lifespan [97].
In C. elegans, TOR deficiency resulted in a more than doubled lifespan [98]. Diminished TOR signaling extends lifespan by promoting autophagy and inhibiting translation, like in yeast [93,99]. Mutation in daf-16 did not abrogate the extended lifespan of LET-363/TOR-(RNAi) worms. However, RNAi directly towards TOR did not further extend the lifespan of long-live daf-2 mutant, suggesting that mTOR may act as a downstream target or independent of daf-16 [98]. daf-15, the C. elegans ortholog of Raptor, was shown to interact with let-363 to regulate metabolism and lifespan [100]. A heterozygous daf-15 mutation led to a 30% increase in mean lifespan and increased fat accumulation, and mutation in daf-16 suppressed longevity effect of daf-15, but did not affect fat accumulation in daf-15 mutant [100]. In addition, daf-16 was found to inhibit daf-15 transcription [100]. Therefore, in C. elegans, the regulation of let-363/daf-15 signaling on lifespan is dependent on daf-16, whereas its metabolic effect is independent of daf-16.
In mice, late-life treatment with rapamycin resulted in an about 10% increase in lifespan [101]. Another study with early-life intervention of rapamycin indicated similar results [102]. Moreover, mice exposed to a DR diet had lower levels of mTOR activity [103]. These findings suggest that mTOR mediates in part mice lifespan extension in response to DR. A study comparing features of rapamycin-treated mice and DR-treated mice demonstrated that rapamycin treatment resulted in an increase in fasting glucose which was not observed in DR animals, without altering fasting insulin, leptin, IGF-1, T4 or FGF-21, which were significantly changed after a five-month DR regimen [104]. It was also reported that rapamycin and DR have quite different effects on transcriptome and metabolome of mice liver, revealing that a combination of these two interventions may produce more benefits than each individual treatment [105]. In addition, a recent study found that although reduced mTOR signaling and DR extended lifespan in rodents, reduced mTOR signaling solely, it did not decrease aging-related mortality, but increased resilience to mortality rate during the aging process [106]. Such distinct mortality patterns have been suggested to reflect differences in insulin sensitivity, mitochondrial biology, xenobiotic metabolism and fatty acid oxidation [106]. To sum up, DR extends lifespan by suppressing TOR activity in a daf-16/FOXO-dependent manner [9]. The findings based on genetic mTOR manipulations and pharmacological treatments indicate that although diminished mTOR signaling has a vital role in DR response, these two longevity-benefiting interventions may act through both distinct and overlapping signaling pathways.

6. Functional Cross Talk Between FOXO-Related Signaling Pathways in DR

The Insulin/IGF-1, AMPK, SIR2, mTOR interact with FOXO to regulate DR response. Besides interacting with FOXO in a single signaling pathway, these signaling modules are serving many interconnected cellular pathways [40]. In the Insulin/IGF pathway, AKT is activated by phosphorylation at Thr-308 by PDK-1, or at Ser-473 by mTORC2 [107]. AKT regulates a variety of downstream effector substrates. AKT can also indirectly activates mTORC1 through TSC1/TSC2 (tuberous sclerosis complex 1/tuberous sclerosis complex 2) and Rheb (Ras homolog enriched in brain) [108]. The activated mTORC1, in turn, inhibits AKT, generating a negative feedback signaling loop [108]. A similar negative feedback mechanism exists between AKT and AMPK. AKT phosphorylates AMPKα1/α2 at Ser-485/491 to attenuate the activation of AMPK by phosphorylation at Thr-172 [109,110]. Reversely, AMPK phosphorylates insulin receptor substrate-1 (IRS-1) at Ser-794, which inhibits PI3K/AKT signaling [111,112]. In addition, AMPK can also upregulate the Akt PH Domain and leucine rich repeat protein phosphatase 2 (PHLPP2) to repress Akt activation [113]. However, on the other hand, AMPK can promote AKT phosphorylation independent of IRS-1 [114,115]. Therefore, the antagonism between AMPK and AKT phosphorylation activities is complex and needs further investigations [116].
It is well-established that AKT and AMPK phosphorylate FOXOs at different amino acids, and result in inactivation or activation, respectively. Interestingly, FOXOs can, in turn, regulate insulin signaling and AMPK activation. Under stress conditions, activated FoxO1 stimulates Akt activation through the suppression of mTORC1 and the elevation of mTORC2 activity [117]. In addition, FOXO can also amplify insulin signaling by upregulating insulin receptor (InsR) mRNA level [118,119]. FOXOs promotes the transcription of Sestrin 3 (a highly conserved, antioxidant, stress-inducible protein that inhibits TORC1 signaling) and led to AMPK activation [120,121].
The interaction between AMPK and Sirtuins is also bidirectional. It was reported that AMPK could indirectly enhance SIRT1 activity by increasing cellular NAD+ levels [122]. On the contrary, SIRT1 deacetylates LKB1 and led to the activation of AMPK [123,124]. SIRT1 was also shown to deacetylate Akt to promote its binding to phosphatidylinositol (3,4,5)-trisphosphate (PIP3) [125]. However, in tumor cells, SIRT1 could also negatively regulate AKT pathway by deacetylation of phosphatase and tensin homolog (PTEN) tumor suppressor protein [126]. In addition, another member of the mammalian Sirtuin family SIRT6 was shown to suppress the expression of Akt signaling-related genes [125]. Therefore, Sirtuins appears to have dual effects on AKT signaling.
The mTOR signaling is also regulated by AMPK and Sirtuins. mTORC1 is negatively regulated by both AMPK and Sirtuins. AMPK can directly phosphorylate tuberous sclerosis complex 2 (TSC2), also known as Tuberin, and Raptor to suppress mTORC1 [127]. mTORC1 is also negatively regulated by SIRT1, while SIRT1 modulates mTORC2 by a SIRT1/mTORC2/Akt signaling pathway [40,128,129]. mTORC2 inhibits the transcriptional activity of FOXO by inducing Akt hyper-activation [130]. FOXO could inhibit mTORC1 activity through activation of TSC1 and increasing mTORC2 activity by elevating Rictor [117,131].
The interactions between FOXOs and co-activators, for example, PGC-1α, also play important roles in DR response [23]. PGC-1α has been shown to interact directly with both FoxO1 and FoxO3 and activate FOXO-dependent gene transcription [132,133]. In addition, FOXOs can bind and stimulate the PGC-1α promoter, representing an auto-regulatory feedback mechanism [133,134]. Also, SIRT1 can interact with and deacetylate PGC-1α to enhance its transcriptional activity [135].

7. Conclusions and Perspectives

The network of DR-related signaling pathways is highly interconnected enabling signaling cross-talk (Figure 1). Although different DR regimens have been shown to lead to the same endpoint, they utilize distinct and overlapped signaling modules to extend lifespan and health-span. Although FOXOs are required for longevity induced by many different DR regimens, it only partially mediates the DR response. One possible hypothesis of such diversity is claiming that different nutrients in the various DR regimens differentially stimulate energy-sensing pathways [62,63]. For example, reduced carbohydrate intake prefers activation of AMPK/FOXO pathway, whereas TOR signaling may mediate responses to restricted amino acid intake [62,136]. Interestingly, an important study in humans demonstrated that diminished protein, but not calorie intake, could decrease circulating IGF-1 levels, suggesting that protein food supply may be a key determinant of the effect of DR interventions [48]. As many studies are focusing on DR methods without malnutrition, further studies aimed to define the specific role of different dietary nutrients in DR are needed. In addition, a better understanding of the precise role of FOXOs in different aspects of DR response would provide a clear insight as to how DR confers lifespan and health-span benefits.
Despite DR has a robust positive impact on several parameters of health and reduces the risk of developing age-related diseases in humans, whether or not DR could extend lifespan in humans remains to be clarified. In addition, DR regimens may be difficult to be implemented and managed in our daily life. Therefore, eliciting DR-related genetic pathways using drugs could be a promising option to achieve DR-like benefits. These potential new drugs defined as calorie restriction mimetics (CRM) could simulate beneficial effects of DR, without limiting food intake. For example, metformin, a biguanide drug commonly used to treat type-2 diabetes, has been shown to extend lifespan and delay the aging process in a variety of species including silkworm Bombyx mori, C. elegans, mice and rats [137,138,139,140]. Although metformin is a potent AMPK activator in all these species, its beneficial action varied. In silkworm B. mori, metformin extends lifespan through AMPK-p53-FOXO pathway [140]. In C. elegans and mice, the longevity-promoting effect of metformin was attributed to the activation of AMPK/Nrf2 pathway [137,139]. Surprisingly, metformin failed to extend lifespan in Drosophila although it activates AMPK and reduces lipid accumulation [141]. Resveratrol, a natural polyphenol antioxidant, has been found to extend lifespan in C. elegans, Drosophila, zebrafish, mice and rats [91,142,143,144]. The longevity benefits of resveratrol are dependent on sirtuins, FOXOs and AMPK [124,142,145]. Several studies demonstrated that resveratrol increased longevity only in obese in mice [146,147]. Rapamycin, an antibiotic targeting mTOR, is also a promising CRM in a variety of organisms [95,101]. It is important to stress that rapamycin could cause adverse effects including diabetic-like symptoms [148]. The precise pharmacological action of these CRMs on DR-related or -independent signaling pathways remains to be further investigated. More efforts should be made in research and development of novel safe CRMs with minimal adverse effects.
In conclusion, the present review summarizes recent research advances in unveiling the molecular mechanisms of FOXOs and related signaling pathways contributions to DR response. Additional experimentation is required to establish a better understanding of the complex signaling network which can be targeted in developing new strategies to extend human longevity. As different DR regimens or DR mimetics can elicit both distinct and overlapping DR-related signaling pathways and may have other actions beyond DR-related mechanisms, it is tempting to propose that the longevity benefits of DR may be maximized by combining diverse forms of DR interventions.

Funding

This research was supported by the National Natural Science Foundation of China (File No. 31771128), The Science and Technology Development Fund, Macau SAR (File No. 0044/2019/AGJ and 0113/2018/A3), The FHS Internal Collaboration Proposals 2017, University of Macau (File No. MYRG2016-00052-FHS and MYRG2018-00134-FHS).

Acknowledgments

Philip Lazarovici holds the Jacob Gitlin Chair in Physiology and is affiliated with, and partially supported by, the Grass Center for Drug Design and Synthesis of Novel Therapeutics, David R. Bloom Center of Pharmacy, and the Adolph and Klara Brettler Medical Research Center at the Hebrew University of Jerusalem, Israel.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sun, D.; Muthukumar, A.R.; Lawrence, R.A.; Fernandes, G. Effects of calorie restriction on polymicrobial peritonitis induced by cecum ligation and puncture in young C57BL/6 mice. Clin. Diagn. Lab. Immunol. 2001, 8, 1003–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Rickenbacher, A.; Jang, J.H.; Limani, P.; Ungethum, U.; Lehmann, K.; Oberkofler, C.E.; Weber, A.; Graf, R.; Humar, B.; Cavien, P.A. Fasting protects liver from ischemic injury through Sirt1-mediated downregulation of circulating HMGB1 in mice. J. Hepatol. 2014, 61, 301–308. [Google Scholar] [CrossRef] [PubMed]
  3. Mitchell, J.R.; Verweij, M.; Brand, K.; van de Ven, M.; Goemaere, N.; van den Engel, S.; Chu, T.; Forrer, F.; Muller, C.; de Jong, M.; et al. Short-term dietary restriction and fasting precondition against ischemia reperfusion injury in mice. Aging Cell 2010, 9, 40–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Miyauchi, T.; Uchida, Y.; Kadono, K.; Hirao, H.; Kawasoe, J.; Watanabe, T.; Ueda, S.; Okajima, H.; Terajima, H.; Uemoto, S. Up-regulation of FOXO1 and reduced inflammation by beta-hydroxybutyric acid are essential diet restriction benefits against liver injury. Proc. Natl. Acad. Sci. USA 2019, 116, 13533–13542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Yu, Z.F.; Mattson, M.P. Dietary restriction and 2-deoxyglucose administration reduce focal ischemic brain damage and improve behavioral outcome: Evidence for a preconditioning mechanism. J. Neurosci. Res. 1999, 57, 830–839. [Google Scholar] [CrossRef]
  6. Mauro, C.R.; Tao, M.; Yu, P.; Trevino-Villerreal, J.H.; Longchamp, A.; Kristal, B.S.; Ozaki, C.K.; Mitchell, J.R. Preoperative dietary restriction reduces intimal hyperplasia and protects from ischemia-reperfusion injury. J. Vasc. Surg. 2016, 63, 500–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Heilbronn, L.K.; de Jonge, L.; Frisard, M.I.; DeLany, J.P.; Larson-Meyer, D.E.; Rood, J.; Nguyen, T.; Martin, C.K.; Volaufova, J.; Most, M.M.; et al. Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals—A randomized controlled trial. Jama J. Am. Med. Assoc. 2006, 295, 1539–1548. [Google Scholar] [CrossRef]
  8. Fontana, L.; Klein, S. Aging, adiposity, and calorie restriction. Jama J. Am. Med. Assoc. 2007, 297, 986–994. [Google Scholar] [CrossRef]
  9. Martins, R.; Lithgow, G.J.; Link, W. Long live FOXO: Unraveling the role of FOXO proteins in aging and longevity. Aging Cell 2016, 15, 196–207. [Google Scholar] [CrossRef]
  10. Furuyama, T.; Nakazawa, T.; Nakano, I.; Mori, N. Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues. Biochem. J. 2000, 349, 629–634. [Google Scholar] [CrossRef]
  11. Hwangbo, D.S.; Gershman, B.; Tu, M.P.; Palmer, M.; Tatar, M. Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature 2004, 429, 562–566. [Google Scholar] [CrossRef] [PubMed]
  12. Sun, X.; Chen, W.D.; Wang, Y.D. DAF-16/FOXO Transcription Factor in Aging and Longevity. Front. Pharmacol 2017, 8, 548. [Google Scholar] [CrossRef] [PubMed]
  13. Accili, D.; Arden, K.C. FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 2004, 117, 421–426. [Google Scholar] [CrossRef] [Green Version]
  14. Henderson, S.T.; Johnson, T.E. daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans. Curr. Biol. 2001, 11, 1975–1980. [Google Scholar] [CrossRef] [Green Version]
  15. Morris, B.J.; Wilicoxa, D.C.; Donlon, T.A.; Willcox, B.J. FOXO3: A Major Gene for Human Longevity—A Mini-Review. Gerontology 2015, 61, 515–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Postnikoff, S.D.; Malo, M.E.; Wong, B.; Harkness, T.A. The yeast forkhead transcription factors fkh1 and fkh2 regulate lifespan and stress response together with the anaphase-promoting complex. PLoS Genet. 2012, 8, e1002583. [Google Scholar] [CrossRef] [Green Version]
  17. Furuyama, T.; Yamashita, H.; Kitayama, K.; Higami, Y.; Shimokawa, I.; Mori, N. Effects of aging and caloric restriction on the gene expression of Foxo1, 3, and 4 (FKHR, FKHRL1, and AFX) in the rat skeletal muscles. Microsc. Res. Tech. 2002, 59, 331–334. [Google Scholar] [CrossRef]
  18. Yamaza, H.; Komatsu, T.; Wakita, S.; Kijogi, C.; Park, S.; Hayashi, H.; Chiba, T.; Mori, R.; Furuyama, T.; Mori, N.; et al. FoxO1 is involved in the antineoplastic effect of calorie restriction. Aging Cell 2010, 9, 372–382. [Google Scholar] [CrossRef] [Green Version]
  19. Edwards, M.G.; Anderson, R.M.; Yuan, M.; Kendziorski, C.M.; Weindruch, R.; Prolla, T.A. Gene expression profiling of aging reveals activation of a p53-mediated transcriptional program. BMC Genom. 2007, 8, 80. [Google Scholar] [CrossRef] [Green Version]
  20. Kim, D.H.; Park, M.H.; Chung, K.W.; Kim, M.J.; Jung, Y.R.; Bae, H.R.; Jang, E.J.; Lee, J.S.; Im, D.S.; Yu, B.P.; et al. The essential role of FoxO6 phosphorylation in aging and calorie restriction. Age 2014, 36, 9679. [Google Scholar] [CrossRef] [Green Version]
  21. Yang, D.; Lian, T.; Tu, J.; Gaur, U.; Mao, X.; Fan, X.; Li, D.; Li, Y.; Yang, M. LncRNA mediated regulation of aging pathways in Drosophila melanogaster during dietary restriction. Aging 2016, 8, 2182–2203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Shimokawa, I.; Komatsu, T.; Hayashi, N.; Kim, S.E.; Kawata, T.; Park, S.; Hayashi, H.; Yamaza, H.; Chiba, T.; Mori, R. The life-extending effect of dietary restriction requires Foxo3 in mice. Aging Cell 2015, 14, 707–709. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, D.H.; Park, M.H.; Ha, S.; Bang, E.J.; Lee, Y.; Lee, A.K.; Lee, J.; Yu, B.P.; Chung, H.Y. Anti-inflammatory action of beta-hydroxybutyrate via modulation of PGC-1alpha and FoxO1, mimicking calorie restriction. Aging 2019, 11, 1283–1304. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, Z.; Isik, M.; Moroz, N.; Steinbaugh, M.J.; Zhang, P.; Blackwell, T.K. Dietary Restriction Extends Lifespan through Metabolic Regulation of Innate Immunity. Cell Metab. 2019, 29, 1192–1205.e8. [Google Scholar] [CrossRef] [PubMed]
  25. Zheng, W.H.; Kar, S.; Quirion, R. Insulin-like growth factor-1-induced phosphorylation of the Forkhead family transcription factor FKHRL1 is mediated by Akt kinase in PC12 cells. J. Biol. Chem. 2000, 275, 39152–39158. [Google Scholar] [CrossRef] [Green Version]
  26. Zheng, W.H.; Kar, S.; Dore, S.; Quirion, R. Insulin-like growth factor-1 (IGF-1): A neuroprotective trophic factor acting via the Akt kinase pathway. Adv. Res. Neurodegener. 2000, 8, 261–272. [Google Scholar]
  27. Zhang, X.; Tang, N.; Hadden, T.J.; Rishi, A.K. Akt, FoxO and regulation of apoptosis. Biochim. Biophys. Acta 2011, 1813, 1978–1986. [Google Scholar] [CrossRef] [Green Version]
  28. Zheng, W.H.; Kar, S.; Quirion, R. Insulin-like growth factor-1-induced phosphorylation of transcription factor FKHRL1 is mediated by phosphatidylinositol 3-kinase/Akt kinase and role of this pathway in insulin-like growth factor-1-induced survival of cultured hippocampal neurons. Mol. Pharmacol. 2002, 62, 225–233. [Google Scholar] [CrossRef]
  29. Matsuzaki, H.; Daitoku, H.; Hatta, M.; Tanaka, K.; Fukamizu, A. Insulin-induced phosphorylation of FKHR (Foxo1) targets to proteasomal degradation. Proc. Natl. Acad. Sci. USA 2003, 100, 11285–11290. [Google Scholar] [CrossRef] [Green Version]
  30. Kenyon, C.; Chang, J.; Gensch, E.; Rudner, A.; Tabtiang, R. A C-Elegans Mutant That Lives Twice as Long as Wild-Type. Nature 1993, 366, 461–464. [Google Scholar] [CrossRef]
  31. Friedman, D.B.; Johnson, T.E. A mutation in the age-1 gene in Caenorhabditis elegans lengthens life and reduces hermaphrodite fertility. Genetics 1988, 118, 75–86. [Google Scholar] [PubMed]
  32. Kenyon, C. The first long-lived mutants: Discovery of the insulin/IGF-1 pathway for ageing. Philos. Trans. R. Soc. Lond. Ser. B 2011, 366, 9–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lakowski, B.; Hekimi, S. The genetics of caloric restriction in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 1998, 95, 13091–13096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Houthoofd, K.; Braeckman, B.P.; Johnson, T.E.; Vanfleteren, J.R. Life extension via dietary restriction is independent of the Ins/IGF-1 signalling pathway in Caenorhabditis elegans. Exp. Gerontol. 2003, 38, 947–954. [Google Scholar] [CrossRef]
  35. Clancy, D.J.; Gems, D.; Hafen, E.; Leevers, S.J.; Partridge, L. Dietary restriction in long-lived dwarf flies. Science 2002, 296, 319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Tu, M.P.; Epstein, D.; Tatar, M. The demography of slow aging in male and female Drosophila mutant for the insulin-receptor substrate homologue chico. Aging Cell 2002, 1, 75–80. [Google Scholar] [CrossRef]
  37. Min, K.J.; Yamamoto, R.; Buch, S.; Pankratz, M.; Tatar, M. Drosophila lifespan control by dietary restriction independent of insulin-like signaling. Aging Cell 2008, 7, 199–206. [Google Scholar] [CrossRef] [Green Version]
  38. Giannakou, M.E.; Goss, M.; Partridge, L. Role of dFOXO in lifespan extension by dietary restriction in Drosophila melanogaster: Not required, but its activity modulates the response. Aging Cell 2008, 7, 187–198. [Google Scholar] [CrossRef]
  39. Berryman, D.E.; Christiansen, J.S.; Johannsson, G.; Thorner, M.O.; Kopchick, J.J. Role of the GH/IGF-1 axis in lifespan and healthspan: Lessons from animal models. Growth Horm. IGF Res. 2008, 18, 455–471. [Google Scholar] [CrossRef] [Green Version]
  40. Pan, H.; Finkel, T. Key proteins and pathways that regulate lifespan. J. Biol. Chem. 2017, 292, 6452–6460. [Google Scholar] [CrossRef] [Green Version]
  41. Greer, K.A.; Hughes, L.M.; Masternak, M.M. Connecting serum IGF-1, body size, and age in the domestic dog. Age 2011, 33, 475–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Taormina, G.; Ferrante, F.; Vieni, S.; Grassi, N.; Russo, A.; Mirisola, M.G. Longevity: Lesson from Model Organisms. Genes 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Parr, T. Insulin exposure and aging theory. Gerontology 1997, 43, 182–200. [Google Scholar] [CrossRef] [PubMed]
  44. Coschigano, K.T.; Clemmons, D.; Bellush, L.L.; Kopchick, J.J. Assessment of growth parameters and life span of GHR/BP gene-disrupted mice. Endocrinology 2000, 141, 2608–2613. [Google Scholar] [CrossRef]
  45. Dunn, S.E.; Kari, F.W.; French, J.; Leininger, J.R.; Travlos, G.; Wilson, R.; Barrett, J.C. Dietary restriction reduces insulin-like growth factor I levels, which modulates apoptosis, cell proliferation, and tumor progression in p53-deficient mice. Cancer Res. 1997, 57, 4667–4672. [Google Scholar] [PubMed]
  46. Lettieri-Barbato, D.; Giovannetti, E.; Aquilano, K. Effects of dietary restriction on adipose mass and biomarkers of healthy aging in human. Aging 2016, 8, 3341–3355. [Google Scholar] [CrossRef] [Green Version]
  47. Redman, L.M.; Veldhuis, J.D.; Rood, J.; Smith, S.R.; Williamson, D.; Ravussin, E.; Team, P.C. The effect of caloric restriction interventions on growth hormone secretion in nonobese men and women. Aging Cell 2010, 9, 32–39. [Google Scholar] [CrossRef]
  48. Fontana, L.; Weiss, E.P.; Villareal, D.T.; Klein, S.; Holloszy, J.O. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell 2008, 7, 681–687. [Google Scholar] [CrossRef] [Green Version]
  49. Page, M.M.; Schuster, E.F.; Mudaliar, M.; Herzyk, P.; Withers, D.J.; Selman, C. Common and unique transcriptional responses to dietary restriction and loss of insulin receptor substrate 1 (IRS1) in mice. Aging-US 2018, 10, 1027–1052. [Google Scholar] [CrossRef]
  50. Zhou, Y.H.; Xu, B.X.C.; Maheshwari, H.G.; He, L.; Reed, M.; Lozykowski, M.; Okada, S.; Cataldo, L.; Coschigamo, K.; Wagner, T.E.; et al. A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc. Natl. Acad. Sci. USA 1997, 94, 13215–13220. [Google Scholar] [CrossRef] [Green Version]
  51. Al-Regaiey, K.A.; Masternak, M.M.; Bonkowski, M.S.; Panici, J.A.; Kopchick, J.J.; Bartke, A. Effects of caloric restriction and growth hormone resistance on insulin-related intermediates in the skeletal muscle. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2007, 62, 18–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Chen, W.Y.; White, M.E.; Wagner, T.E.; Kopchick, J.J. Functional Antagonism Between Endogenous Mouse Growth-Hormone (Gh) And a Gh Analog Results In Dwarf Transgenic Mice. Endocrinology 1991, 129, 1402–1408. [Google Scholar] [CrossRef] [PubMed]
  53. Berryman, D.E.; Lubbers, E.R.; Magon, V.; List, E.O.; Kopchick, J.J. A dwarf mouse model with decreased GH/IGF-1 activity that does not experience life-span extension: Potential impact of increased adiposity, leptin, and insulin with advancing age. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 131–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bartke, A.; Masternak, M.M.; Al-Regaiey, K.A.; Bonkowski, M.S. Effects of dietary restriction on the expression of insulin-signaling-related genes in long-lived mutant mice. Interdiscip. Top. Gerontol. 2007, 35, 69–82. [Google Scholar] [CrossRef] [PubMed]
  55. Westbrook, R.; Bonkowski, M.S.; Arum, O.; Strader, A.D.; Bartke, A. Metabolic Alterations Due to Caloric Restriction and Every Other Day Feeding in Normal and Growth Hormone Receptor Knockout Mice. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2014, 69, 25–33. [Google Scholar] [CrossRef] [Green Version]
  56. Long, Y.C.; Zierath, J.R. AMP-activated protein kinase signaling in metabolic regulation. J. Clin. Investig. 2006, 116, 1776–1783. [Google Scholar] [CrossRef] [Green Version]
  57. Greer, E.L.; Oskoui, P.R.; Banko, M.R.; Maniar, J.M.; Gygi, M.P.; Gygi, S.P.; Brunet, A. The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J. Biol. Chem. 2007, 282, 30107–30119. [Google Scholar] [CrossRef] [Green Version]
  58. Apfeld, J.; O′Connor, G.; McDonagh, T.; DiStefano, P.S.; Curtis, R. The AMP-activated protein kinase AAK-2 links energy levels and insulin-like signals to lifespan in C. elegans. Genes Dev. 2004, 18, 3004–3009. [Google Scholar] [CrossRef] [Green Version]
  59. Houthoofd, K.; Johnson, T.E.; Vanfleteren, J.R. Dietary restriction in the nematode Caenorhabditis elegans. J. Gerontol. A Biol. Sci. Med. Sci. 2005, 60, 1125–1131. [Google Scholar] [CrossRef] [Green Version]
  60. Curtis, R.; O′Connor, G.; DiStefano, P.S. Aging networks in Caenorhabditis elegans: AMP-activated protein kinase (aak-2) links multiple aging and metabolism pathways. Aging Cell 2006, 5, 119–126. [Google Scholar] [CrossRef]
  61. Greer, E.L.; Dowlatshahi, D.; Banko, M.R.; Villen, J.; Hoang, K.; Blanchard, D.; Gygi, S.P.; Brunet, A. An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr. Biol. 2007, 17, 1646–1656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Greer, E.L.; Brunet, A. Different dietary restriction regimens extend lifespan by both independent and overlapping genetic pathways in C. elegans. Aging Cell 2009, 8, 113–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Greer, E.L.; Banko, M.R.; Brunet, A. AMP-activated protein kinase and FoxO transcription factors in dietary restriction-induced longevity. Ann. N. Y. Acad. Sci. 2009, 1170, 688–692. [Google Scholar] [CrossRef] [PubMed]
  64. Frye, R.A. Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem. Biophys. Res. Commun. 2000, 273, 793–798. [Google Scholar] [CrossRef]
  65. Schiedel, M.; Robaa, D.; Rumpf, T.; Sippl, W.; Jung, M. The Current State of NAD(+)-Dependent Histone Deacetylases (Sirtuins) as Novel Therapeutic Targets. Med. Res. Rev. 2018, 38, 147–200. [Google Scholar] [CrossRef]
  66. Tissenbaum, H.A.; Guarente, L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature 2001, 410, 227–230. [Google Scholar] [CrossRef]
  67. Rogina, B.; Helfand, S.L. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc. Natl. Acad. Sci. USA 2004, 101, 15998–16003. [Google Scholar] [CrossRef] [Green Version]
  68. Daitoku, H.; Sakamaki, J.; Fukamizu, A. Regulation of FoxO transcription factors by acetylation and protein-protein interactions. Biochim. Biophys. Acta 2011, 1813, 1954–1960. [Google Scholar] [CrossRef] [Green Version]
  69. Matsuzaki, H.; Daitoku, H.; Hatta, M.; Aoyama, H.; Yoshimochi, K.; Fukamizu, A. Acetylation of Foxo1 alters its DNA-binding ability and sensitivity to phosphorylation. Proc. Natl. Acad. Sci. USA 2005, 102, 11278–11283. [Google Scholar] [CrossRef] [Green Version]
  70. Brunet, A.; Sweeney, L.B.; Sturgill, J.F.; Chua, K.F.; Greer, P.L.; Lin, Y.; Tran, H.; Ross, S.E.; Mostoslavsky, R.; Cohen, H.Y.; et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004, 303, 2011–2015. [Google Scholar] [CrossRef] [Green Version]
  71. Frescas, D.; Valenti, L.; Accili, D. Nuclear trapping of the forkhead transcription factor FoxO1 via Sirt-dependent deacetylation promotes expression of glucogenetic genes. J. Biol. Chem. 2005, 280, 20589–20595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Wang, F.; Nguyen, M.; Qin, F.X.; Tong, Q. SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell 2007, 6, 505–514. [Google Scholar] [CrossRef] [PubMed]
  73. Bamps, S.; Wirtz, J.; Savory, F.R.; Lake, D.; Hope, I.A. The Caenorhabditis elegans sirtuin gene, sir-2.1, is widely expressed and induced upon caloric restriction. Mech. Ageing Dev. 2009, 130, 762–770. [Google Scholar] [CrossRef] [PubMed]
  74. Lin, S.J.; Defossez, P.A.; Guarente, L. Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science 2000, 289, 2126–2128. [Google Scholar] [CrossRef] [Green Version]
  75. Berdichevsky, A.; Viswanathan, M.; Horvitz, H.R.; Guarente, L. C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate DAF-16 and extend life span. Cell 2006, 125, 1165–1177. [Google Scholar] [CrossRef] [Green Version]
  76. Uno, M.; Nishida, E. Lifespan-regulating genes in C. elegans. NPJ Aging Mech Dis 2016, 2, 16010. [Google Scholar] [CrossRef]
  77. Wang, Y.; Tissenbaum, H.A. Overlapping and distinct functions for a Caenorhabditis elegans SIR2 and DAF-16/FOXO. Mech. Ageing Dev. 2006, 127, 48–56. [Google Scholar] [CrossRef]
  78. Kaeberlein, M.; Kirkland, K.T.; Fields, S.; Kennedy, B.K. Sir2-independent life span extension by calorie restriction in yeast. PLoS Biol. 2004, 2, E296. [Google Scholar] [CrossRef]
  79. Tsuchiya, M.; Dang, N.; Kerr, E.O.; Hu, D.; Steffen, K.K.; Oakes, J.A.; Kennedy, B.K.; Kaeberlein, M. Sirtuin-independent effects of nicotinamide on lifespan extension from calorie restriction in yeast. Aging Cell 2006, 5, 505–514. [Google Scholar] [CrossRef]
  80. Braidy, N.; Guillemin, G.J.; Mansour, H.; Chan-Ling, T.; Poljak, A.; Grant, R. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS ONE 2011, 6, e19194. [Google Scholar] [CrossRef]
  81. Quintas, A.; de Solis, A.J.; Diez-Guerra, F.J.; Carrascosa, J.M.; Bogonez, E. Age-associated decrease of SIRT1 expression in rat hippocampus: Prevention by late onset caloric restriction. Exp. Gerontol. 2012, 47, 198–201. [Google Scholar] [CrossRef] [PubMed]
  82. Cohen, H.Y.; Miller, C.; Bitterman, K.J.; Wall, N.R.; Hekking, B.; Kessler, B.; Howitz, K.T.; Gorospe, M.; de Cabo, R.; Sinclair, D.A. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 2004, 305, 390–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lee, S.M.; Yang, H.; Tartar, D.M.; Gao, B.; Luo, X.; Ye, S.Q.; Zaghouani, H.; Fang, D. Prevention and treatment of diabetes with resveratrol in a non-obese mouse model of type 1 diabetes. Diabetologia 2011, 54, 1136–1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chang, J.; Rimando, A.; Pallas, M.; Camins, A.; Porquet, D.; Reeves, J.; Shukitt-Hale, B.; Smith, M.A.; Joseph, J.A.; Casadesus, G. Low-dose pterostilbene, but not resveratrol, is a potent neuromodulator in aging and Alzheimer’s disease. Neurobiol. Aging 2012, 33, 2062–2071. [Google Scholar] [CrossRef]
  85. Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11. [Google Scholar] [CrossRef] [Green Version]
  86. Kimura, Y.; Sumiyoshi, M. Resveratrol Prevents Tumor Growth and Metastasis by Inhibiting Lymphangiogenesis and M2 Macrophage Activation and Differentiation in Tumor-associated Macrophages. Nutr. Cancer 2016, 68, 667–678. [Google Scholar] [CrossRef]
  87. Nawaz, W.; Zhou, Z.; Deng, S.; Ma, X.; Ma, X.; Li, C.; Shu, X. Therapeutic Versatility of Resveratrol Derivatives. Nutrients 2017, 9. [Google Scholar] [CrossRef] [Green Version]
  88. Elibol, B.; Kilic, U. High Levels of SIRT1 Expression as a Protective Mechanism Against Disease-Related Conditions. Front. Endocrinol. 2018, 9. [Google Scholar] [CrossRef]
  89. Fang, E.F.; Lautrup, S.; Hou, Y.; Demarest, T.G.; Croteau, D.L.; Mattson, M.P.; Bohr, V.A. NAD(+) in Aging: Molecular Mechanisms and Translational Implications. Trends Mol. Med. 2017, 23, 899–916. [Google Scholar] [CrossRef]
  90. Chen, D.; Steele, A.D.; Lindquist, S.; Guarente, L. Increase in activity during calorie restriction requires Sirt1. Science 2005, 310, 1641. [Google Scholar] [CrossRef] [Green Version]
  91. Wood, J.G.; Rogina, B.; Lavu, S.; Howitz, K.; Helfand, S.L.; Tatar, M.; Sinclair, D. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 2004, 430, 686–689. [Google Scholar] [CrossRef] [PubMed]
  92. Kennedy, B.K.; Lamming, D.W. The Mechanistic Target of Rapamycin: The Grand ConducTOR of Metabolism and Aging. Cell Metab. 2016, 23, 990–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hansen, M.; Taubert, S.; Crawford, D.; Libina, N.; Lee, S.J.; Kenyon, C. Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans. Aging Cell 2007, 6, 95–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Boutouja, F.; Stiehm, C.M.; Platta, H.W. mTOR: A Cellular Regulator Interface in Health and Disease. Cells 2019, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Powers, R.W., 3rd; Kaeberlein, M.; Caldwell, S.D.; Kennedy, B.K.; Fields, S. Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes Dev. 2006, 20, 174–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kaeberlein, M.; Powers, R.W., 3rd; Steffen, K.K.; Westman, E.A.; Hu, D.; Dang, N.; Kerr, E.O.; Kirkland, K.T.; Fields, S.; Kennedy, B.K. Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients. Science 2005, 310, 1193–1196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kapahi, P.; Zid, B.M.; Harper, T.; Koslover, D.; Sapin, V.; Benzer, S. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr. Biol. 2004, 14, 885–890. [Google Scholar] [CrossRef] [Green Version]
  98. Vellai, T.; Takacs-Vellai, K.; Zhang, Y.; Kovacs, A.L.; Orosz, L.; Muller, F. Genetics: Influence of TOR kinase on lifespan in C. elegans. Nature 2003, 426, 620. [Google Scholar] [CrossRef]
  99. Hansen, M.; Chandra, A.; Mitic, L.L.; Onken, B.; Driscoll, M.; Kenyon, C. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS Genet. 2008, 4, e24. [Google Scholar] [CrossRef] [Green Version]
  100. Jia, K.; Chen, D.; Riddle, D.L. The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development 2004, 131, 3897–3906. [Google Scholar] [CrossRef] [Green Version]
  101. Harrison, D.E.; Strong, R.; Sharp, Z.D.; Nelson, J.F.; Astle, C.M.; Flurkey, K.; Nadon, N.L.; Wilkinson, J.E.; Frenkel, K.; Carter, C.S.; et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 2009, 460, 392–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Miller, R.A.; Harrison, D.E.; Astle, C.M.; Baur, J.A.; Boyd, A.R.; de Cabo, R.; Fernandez, E.; Flurkey, K.; Javors, M.A.; Nelson, J.F.; et al. Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J. Gerontol. A Biol. Sci. Med. Sci. 2011, 66, 191–201. [Google Scholar] [CrossRef] [PubMed]
  103. Sun, L.; Akha, A.A.S.; Miller, R.A.; Harper, J.M. Life-Span Extension in Mice by Preweaning Food Restriction and by Methionine Restriction in Middle Age. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2009, 64, 711–722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Miller, R.A.; Harrison, D.E.; Astle, C.M.; Fernandez, E.; Flurkey, K.; Han, M.; Javors, M.A.; Li, X.N.; Nadon, N.L.; Nelson, J.F.; et al. Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell 2014, 13, 468–477. [Google Scholar] [CrossRef] [PubMed]
  105. Fok, W.C.; Bokov, A.; Gelfond, J.; Yu, Z.; Zhang, Y.Q.; Doderer, M.; Chen, Y.D.; Javors, M.; Wood, W.H.; Zhang, Y.Q.; et al. Combined treatment of rapamycin and dietary restriction has a larger effect on the transcriptome and metabolome of liver. Aging Cell 2014, 13, 311–319. [Google Scholar] [CrossRef]
  106. Garratt, M.; Nakagawa, S.; Simons, M.J. Comparative idiosyncrasies in life extension by reduced mTOR signalling and its distinctiveness from dietary restriction. Aging Cell 2016, 15, 737–743. [Google Scholar] [CrossRef] [Green Version]
  107. Hemmings, B.A.; Restuccia, D.F. PI3K-PKB/Akt pathway. Cold Spring Harb. Perspect. Biol. 2012, 4, a011189. [Google Scholar] [CrossRef] [Green Version]
  108. Bhaskar, P.T.; Hay, N. The two TORCs and Akt. Dev. Cell 2007, 12, 487–502. [Google Scholar] [CrossRef] [Green Version]
  109. Kovacic, S.; Soltys, C.L.; Barr, A.J.; Shiojima, I.; Walsh, K.; Dyck, J.R. Akt activity negatively regulates phosphorylation of AMP-activated protein kinase in the heart. J. Biol. Chem. 2003, 278, 39422–39427. [Google Scholar] [CrossRef] [Green Version]
  110. Zaha, V.G.; Young, L.H. AMP-activated protein kinase regulation and biological actions in the heart. Circ. Res. 2012, 111, 800–814. [Google Scholar] [CrossRef] [Green Version]
  111. Tzatsos, A.; Tsichlis, P.N. Energy depletion inhibits phosphatidylinositol 3-kinase/Akt signaling and induces apoptosis via AMP-activated protein kinase-dependent phosphorylation of IRS-1 at Ser-794. J. Biol. Chem. 2007, 282, 18069–18082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Ning, J.; Clemmons, D.R. AMP-activated protein kinase inhibits IGF-I signaling and protein synthesis in vascular smooth muscle cells via stimulation of insulin receptor substrate 1 S794 and tuberous sclerosis 2 S1345 phosphorylation. Mol. Endocrinol. 2010, 24, 1218–1229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Saha, M.; Kumar, S.; Bukhari, S.; Balaji, S.A.; Kumar, P.; Hindupur, S.K.; Rangarajan, A. AMPK-Akt Double-Negative Feedback Loop in Breast Cancer Cells Regulates Their Adaptation to Matrix Deprivation. Cancer Res. 2018, 78, 1497–1510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Tao, R.; Gong, J.; Luo, X.; Zang, M.; Guo, W.; Wen, R.; Luo, Z. AMPK exerts dual regulatory effects on the PI3K pathway. J. Mol. Signal. 2010, 5, 1. [Google Scholar] [CrossRef] [Green Version]
  115. Han, F.; Li, C.F.; Cai, Z.; Zhang, X.; Jin, G.; Zhang, W.N.; Xu, C.; Wang, C.Y.; Morrow, J.; Zhang, S.; et al. The critical role of AMPK in driving Akt activation under stress, tumorigenesis and drug resistance. Nat. Commun. 2018, 9, 4728. [Google Scholar] [CrossRef]
  116. Zhao, Y.; Hu, X.B.; Liu, Y.J.; Dong, S.M.; Wen, Z.W.; He, W.M.; Zhang, S.Y.; Huang, Q.; Shi, M. ROS signaling under metabolic stress: Cross-talk between AMPK and AKT pathway. Mol. Cancer 2017, 16. [Google Scholar] [CrossRef] [Green Version]
  117. Chen, C.C.; Jeon, S.M.; Bhaskar, P.T.; Nogueira, V.; Sundararajan, D.; Tonic, I.; Park, Y.; Hay, N. FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3 and Rictor. Dev. Cell 2010, 18, 592–604. [Google Scholar] [CrossRef] [Green Version]
  118. Marr, M.T., 2nd; D′Alessio, J.A.; Puig, O.; Tjian, R. IRES-mediated functional coupling of transcription and translation amplifies insulin receptor feedback. Genes Dev. 2007, 21, 175–183. [Google Scholar] [CrossRef] [Green Version]
  119. Hay, N. Interplay between FOXO, TOR, and Akt. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 1965–1970. [Google Scholar] [CrossRef] [Green Version]
  120. Lee, J.H.; Budanov, A.V.; Karin, M. Sestrins orchestrate cellular metabolism to attenuate aging. Cell Metab. 2013, 18, 792–801. [Google Scholar] [CrossRef] [Green Version]
  121. Hong-Brown, L.Q.; Brown, C.R.; Navaratnarajah, M.; Lang, C.H. Adamts1 mediates ethanol-induced alterations in collagen and elastin via a FoxO1-sestrin3-AMPK signaling cascade in myocytes. J. Cell. Biochem. 2015, 116, 91–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Canto, C.; Gerhart-Hines, Z.; Feige, J.N.; Lagouge, M.; Noriega, L.; Milne, J.C.; Elliott, P.J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 2009, 458, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  123. Hou, X.Y.; Xu, S.Q.; Maitland-Toolan, K.A.; Sato, K.; Jiang, B.B.; Ido, Y.S.; Lan, F.; Walsh, K.; Wierzbicki, M.; Verbeuren, T.J.; et al. SIRT1 regulates hepatocyte lipid metabolism through activating AMP-activated protein kinase. J. Biol. Chem. 2008, 283, 20015–20026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Price, N.L.; Gomes, A.P.; Ling, A.J.Y.; Duarte, F.V.; Martin-Montalvo, A.; North, B.J.; Agarwal, B.; Ye, L.; Ramadori, G.; Teodoro, J.S.; et al. SIRT1 Is Required for AMPK Activation and the Beneficial Effects of Resveratrol on Mitochondrial Function. Cell Metab. 2012, 15, 675–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Pillai, V.B.; Sundaresan, N.R.; Gupta, M.P. Regulation of Akt Signaling by Sirtuins Its Implication in Cardiac Hypertrophy and Aging. Circ. Res. 2014, 114, 368–378. [Google Scholar] [CrossRef] [Green Version]
  126. Chai, R.F.; Fu, H.L.; Zheng, Z.D.; Liu, T.T.; Ji, S.H.; Li, G.R. Resveratrol inhibits proliferation and migration through SIRT1 mediated post-translational modification of PI3K/AKT signaling in hepatocellular carcinoma cells. Mol. Med. Rep. 2017, 16, 8037–8044. [Google Scholar] [CrossRef] [Green Version]
  127. Shaw, R.J. LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol. 2009, 196, 65–80. [Google Scholar] [CrossRef]
  128. Ghosh, H.S.; McBurney, M.; Robbins, P.D. SIRT1 negatively regulates the mammalian target of rapamycin. PLoS ONE 2010, 5, e9199. [Google Scholar] [CrossRef] [Green Version]
  129. Wang, R.H.; Kim, H.S.; Xiao, C.; Xu, X.; Gavrilova, O.; Deng, C.X. Hepatic Sirt1 deficiency in mice impairs mTorc2/Akt signaling and results in hyperglycemia, oxidative damage, and insulin resistance. J. Clin. Investig. 2011, 121, 4477–4490. [Google Scholar] [CrossRef] [Green Version]
  130. Mori, S.; Nada, S.; Kimura, H.; Tajima, S.; Takahashi, Y.; Kitamura, A.; Oneyama, C.; Okada, M. The mTOR pathway controls cell proliferation by regulating the FoxO3a transcription factor via SGK1 kinase. PLoS ONE 2014, 9, e88891. [Google Scholar] [CrossRef] [Green Version]
  131. Khatri, S.; Yepiskoposyan, H.; Gallo, C.A.; Tandon, P.; Plas, D.R. FOXO3a Regulates Glycolysis via Transcriptional Control of Tumor Suppressor TSC1. J. Biol. Chem. 2010, 285, 15960–15965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Puigserver, P.; Rhee, J.; Donovan, J.; Walkey, C.J.; Yoon, J.C.; Oriente, F.; Kitamura, Y.; Altomonte, J.; Dong, H.; Accili, D.; et al. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. Nature 2003, 423, 550–555. [Google Scholar] [CrossRef] [PubMed]
  133. Olmos, Y.; Valle, I.; Borniquel, S.; Tierrez, A.; Soria, E.; Lamas, S.; Monsalve, M. Mutual dependence of Foxo3a and PGC-1alpha in the induction of oxidative stress genes. J. Biol. Chem. 2009, 284, 14476–14484. [Google Scholar] [CrossRef] [Green Version]
  134. Fernandez-Marcos, P.J.; Auwerx, J. Regulation of PGC-1alpha, a nodal regulator of mitochondrial biogenesis. Am. J. Clin. Nutr. 2011, 93, 884S–890S. [Google Scholar] [CrossRef] [Green Version]
  135. Rodgers, J.T.; Lerin, C.; Haas, W.; Gygi, S.P.; Spiegelman, B.M.; Puigserver, P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 2005, 434, 113–118. [Google Scholar] [CrossRef]
  136. Avruch, J.; Hara, K.; Lin, Y.; Liu, M.; Long, X.; Ortiz-Vega, S.; Yonezawa, K. Insulin and amino-acid regulation of mTOR signaling and kinase activity through the Rheb GTPase. Oncogene 2006, 25, 6361–6372. [Google Scholar] [CrossRef] [Green Version]
  137. Onken, B.; Driscoll, M. Metformin induces a dietary restriction-like state and the oxidative stress response to extend C. elegans Healthspan via AMPK, LKB1, and SKN-1. PLoS ONE 2010, 5, e8758. [Google Scholar] [CrossRef]
  138. Smith, D.L., Jr.; Elam, C.F., Jr.; Mattison, J.A.; Lane, M.A.; Roth, G.S.; Ingram, D.K.; Allison, D.B. Metformin supplementation and life span in Fischer-344 rats. J. Gerontol. A Biol. Sci. Med. Sci. 2010, 65, 468–474. [Google Scholar] [CrossRef] [Green Version]
  139. Martin-Montalvo, A.; Mercken, E.M.; Mitchell, S.J.; Palacios, H.H.; Mote, P.L.; Scheibye-Knudsen, M.; Gomes, A.P.; Ward, T.M.; Minor, R.K.; Blouin, M.J.; et al. Metformin improves healthspan and lifespan in mice. Nat. Commun. 2013, 4, 2192. [Google Scholar] [CrossRef]
  140. Song, J.B.; Jiang, G.H.; Zhang, J.F.; Guo, J.S.; Li, Z.; Hao, K.G.; Liu, L.; Cheng, Z.L.; Tong, X.L.; Dai, F.Y. Metformin prolongs lifespan through remodeling the energy distribution strategy in silkworm, Bombyx mori. Aging-US 2019, 11, 240–248. [Google Scholar] [CrossRef]
  141. Slack, C.; Foley, A.; Partridge, L. Activation of AMPK by the Putative Dietary Restriction Mimetic Metformin Is Insufficient to Extend Lifespan in Drosophila. PLoS ONE 2012, 7. [Google Scholar] [CrossRef] [PubMed]
  142. Bhullar, K.S.; Hubbard, B.P. Lifespan and healthspan extension by resveratrol. Biochim. Biophys. Acta 2015, 1852, 1209–1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Lee, J.; Kwon, G.; Park, J.; Kim, J.K.; Lim, Y.H. Brief Communication: SIR-2.1-dependent lifespan extension of Caenorhabditis elegans by oxyresveratrol and resveratrol. Exp. Biol. Med. (Maywood) 2016, 241, 1757–1763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ding, S.; Jiang, J.; Zhang, G.; Bu, Y.; Zhang, G.; Zhao, X. Resveratrol and caloric restriction prevent hepatic steatosis by regulating SIRT1-autophagy pathway and alleviating endoplasmic reticulum stress in high-fat diet-fed rats. PLoS ONE 2017, 12, e0183541. [Google Scholar] [CrossRef] [Green Version]
  145. Sin, T.K.; Yung, B.Y.; Siu, P.M. Modulation of SIRT1-Foxo1 signaling axis by resveratrol: Implications in skeletal muscle aging and insulin resistance. Cell. Physiol. Biochem. 2015, 35, 541–552. [Google Scholar] [CrossRef]
  146. Pearson, K.J.; Baur, J.A.; Lewis, K.N.; Peshkin, L.; Price, N.L.; Labinskyy, N.; Swindell, W.R.; Kamara, D.; Minor, R.K.; Perez, E.; et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008, 8, 157–168. [Google Scholar] [CrossRef] [Green Version]
  147. Baur, J.A.; Pearson, K.J.; Price, N.L.; Jamieson, H.A.; Lerin, C.; Kalra, A.; Prabhu, V.V.; Allard, J.S.; Lopez-Lluch, G.; Lewis, K.; et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006, 444, 337–342. [Google Scholar] [CrossRef]
  148. Blattler, S.M.; Cunningham, J.T.; Verdeguer, F.; Chim, H.; Haas, W.; Liu, H.F.; Romanino, K.; Ruegg, M.A.; Gygi, S.P.; Shi, Y.; et al. Yin Yang 1 Deficiency in Skeletal Muscle Protects against Rapamycin-Induced Diabetic-like Symptoms through Activation of Insulin/IGF Signaling. Cell Metab. 2012, 15, 505–517. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The signaling network of forkhead box O transcription factors (FOXO)-related pathways in dietary restriction (DR).
Figure 1. The signaling network of forkhead box O transcription factors (FOXO)-related pathways in dietary restriction (DR).
Cells 09 00100 g001

Share and Cite

MDPI and ACS Style

Jiang, Y.; Yan, F.; Feng, Z.; Lazarovici, P.; Zheng, W. Signaling Network of Forkhead Family of Transcription Factors (FOXO) in Dietary Restriction. Cells 2020, 9, 100. https://doi.org/10.3390/cells9010100

AMA Style

Jiang Y, Yan F, Feng Z, Lazarovici P, Zheng W. Signaling Network of Forkhead Family of Transcription Factors (FOXO) in Dietary Restriction. Cells. 2020; 9(1):100. https://doi.org/10.3390/cells9010100

Chicago/Turabian Style

Jiang, Yizhou, Fengxia Yan, Zhongping Feng, Philip Lazarovici, and Wenhua Zheng. 2020. "Signaling Network of Forkhead Family of Transcription Factors (FOXO) in Dietary Restriction" Cells 9, no. 1: 100. https://doi.org/10.3390/cells9010100

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop